• Log in with Facebook Log in with Twitter Log In with Google      Sign In    
  • Create Account
  LongeCity
              Advocacy & Research for Unlimited Lifespans

Photo
* * * * - 6 votes

Alternative methods to extend telomeres

telomeres nad nampt ampk resveratrol allicin methylene blue nmn sirtuins statin

  • Please log in to reply
989 replies to this topic

#151 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 06 December 2019 - 03:51 PM

  Ive looked at Janic studies, there is no mention of it and in one of those LDL-C was lowered little below 20% too.

Having said that I think it would be sensible to go next time for a short half life statin like fluvastatin that Janik have used. Unfortunately they are hard to come by as all modern statins are long lasting therefore more effective.

 

what about using Pravastatin ?

it is the only statin that does not pass brain blood barrier...

also the only one that goes into liver by active transport as I know....


  • like x 1

#152 Andey

  • Guest
  • 673 posts
  • 203
  • Location:Kiev, Ukraine

Posted 06 December 2019 - 05:40 PM

what about using Pravastatin ?

it is the only statin that does not pass brain blood barrier...

also the only one that goes into liver by active transport as I know....

 

  Agree, I noticed pravastatin comes often as one of the safest and non proportionally effective(to its cholesterol lowering) statins.

Unfortunately it doesn't available where I live and arranging getting it from abroad looks like too much hassle.



#153 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 06 December 2019 - 06:54 PM

Agree, I noticed pravastatin comes often as one of the safest and non proportionally effective(to its cholesterol lowering) statins.
Unfortunately it doesn't available where I live and arranging getting it from abroad looks like too much hassle.


It would be interesting to try a hydrophilic statin, but on balance I probably prefer to stay with a lipophilic statin like atorvastatin or simvastatin because it will penetrate cells and the body in general more completely (notwithstanding some hydrophilic molecules having active transporters). Interestingly fluvastatin is an intermediate molecule with both hydo and lipo philic properties.

I use atorvastatin for the oral route and simvastatin in a skin cream due to its lower molecular mass. I have had good results from the oral route but the skin cream has not been successful, other than the fact it heals cuts very quickly. Because atorvastatin is so potent,I only take 5mg/day for the 20 day duration of the protocol.
  • like x 1

sponsored ad

  • Advert

#154 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 07 December 2019 - 06:44 AM

I see problematic killing brain cells even if sensesent...

#155 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 07 December 2019 - 08:35 AM

I thought I'd write this post to explain a little bit more about my view of aging.

I look at aging in a very simple way. Cells are like little powerplants that make their own energy and manufacture the molecules they need. They also clean up the waste this produces, which includes the molecules they've made. But like any waste disposal process, it isn't perfect. Hence to ensure continual (immortal) health a cell has to divide to keep levels of metabolic waste from reaching critical levels.

This ability of dividing cells is one reason bacteria generally don't age, children are healthier than adults, why some animals that stay in a neonatal (child-like)state (i.e. my avatar the axolotl) appear not to age, why animals that grow all their lives appear not to age, and why aging seems to start as soon as growth ceases and adulthood starts.

If cell division ceases then the cell has only cell cleaning to keep it healthy, and this is where autophagy comes in. As soon as autophagy gets behind we get cellular growth (rather than division) and hyperfunction (followed inevitably by dysfunction). Old cells get bigger.

It is interesting that the smaller a cell is, the more stem-like. Pluripotent or embryonic cells are the smallest. Among mesenchymal stem stems, the smaller mesenchymal stem cells are the most totipotent.

So let's talk about telomeres. Telomeres are what is needed to keep cell division going, and to keep harmful metabolic aggregates below a certain level, and to keep cells small. In cells that don't divide by design, or divide very slowly (neurons and cardiomyocyte cells come to mind) there are always other dividing cells on which those non dividing cells depend (glial and astrocytes for neurons, endothelial cells for cardiomyocytes, satellite cells for the muscle, etc.) and which in some cases (glial and astrocytes for neurons) are actually responsible for helping clear the waste (amyloid beta and tau for example).

Additionally, gene expression changes as telomeres shorten,and not only does this slow cell division, but it also slows all protein turnover within the cell. Hence autophagy is failing as a direct result of telomere loss in dividing cells, in addition to the waste disposal problem downstream in some non-dividing cells.

So as we age the cells of the body are getting less numerous, bigger, and less stem-like (and this is probably where methylation changes come in).

Now stem cells spawn progenitor cells, and progenitor cells do much of the dividing that produces somatic cell lines. We get more division in tissues that need more replacement, i.e. more cell division in blood than muscle, for example, so telomeres are shorter in blood than muscle. But telomere rate of loss is about the same for all tissues as we age (https://www.nature.c...cles/ncomms2602). Which tells us that there are less progenitor cells in say muscle than blood (less cells divide less often equals same loss). Hence tissue maintenance is failing across the whole body with age as a result of shortening telomeres, and also leads to a metabolic waste problem in the cells that remain.

So what can we do as a present day intervention?

First things first. As we get older autophagy upregulators are a necessity (some combination of glucosamine, coffee, rapamycin,fasting, exercise, etc.) to keep cells smaller and cleaner until such as time as the telomere problem is addressed.

Secondly, telomeres. We need powerful telomerase activators. This thread has gone some way towards addressing this.

Thirdly, loss of stemness and epigenetic aging. This thread goes some way to addressing this by shrinking cells using ROCK inhibitors and this also contributes their proliferative ability.

I also have a senolytic/anti cancer protocol in preparation(https://www.ncbi.nlm...les/PMC6520007/) which I've mentioned elsewhere, using liposomal Vit C, and two synergistic antibiotics. I will post on this when I have tried it. Another member will also be trialling this. Senolytics are useful in that they can clear out death resistant cells that have become permanently senescent and can't be rejuvenated by the above methods, and also reverse some of the downstream harmful metabolic effects we talked about. But I wouldn't use them as a monotherapy due to the increased requirement for more cell division, which might exacerbate the problem in the long run.

 

The selfish tendency of lazy old cells that tend survive in the body LONGER and MORE than efficient young cells mirrors somehow the selfish gene that regulates them into DNA (and as Richard Dawkins teaches us the tendency toward material stability si really what is deeply selfish) .... beeing DNA the same in the lazy old ones and into the young brave generous cells some kind of epigenetic change should differentiate old and young cells... or what ?
In your view can you explain this ?

https://www.scienced...047637408000961


 


  • Informative x 1

#156 Andey

  • Guest
  • 673 posts
  • 203
  • Location:Kiev, Ukraine

Posted 07 December 2019 - 08:51 AM

 

I am not QuestForLife but abstract for it is mindbogling

Dietary restriction feeding extends survival in a range of species but a detailed understanding of the underlying mechanism is lacking. There is interest therefore in identifying a more targeted approach to replicate this effect on survival. We report that in rats dietary supplementation with alpha-lipoic acid, has markedly differing effects on lifetime survival depending upon the dietary history of the animal. When animals are switched from DR feeding to ad libitum feeding with a diet supplemented with alpha-lipoic acid, the extended survival characteristic of DR feeding is maintained, even though the animals show accelerated growth. Conversely, switching from ad libitum feeding a diet supplemented with alpha-lipoic acid to DR feeding of the non-supplemented diet, blocks the normal effect of DR to extend survival, even after cessation of lipoic acid supplementation. Unlike the dynamic effect of switching between DR and ad libitum feeding with a non-supplemented diet where the subsequent survival trajectory is determined by the new feeding regime, lipoic acid fixes the survival trajectory to that established by the initial feeding regime. Ad libitum feeding a diet supplemented with lipoic acid can therefore act as mimetic of DR to extend survival.

 

 TBH I would like somebody to replicate this first, before thinking too much about how does it work this way.


  • Informative x 1

#157 Oakman

  • Location:CO

Posted 07 December 2019 - 03:11 PM

"When animals are switched from DR feeding to ad libitum feeding with a diet supplemented with alpha-lipoic acid, the extended survival characteristic of DR feeding is maintained".

 

That's a pretty significant result attributed to ALA, and would be amazing if it were proved again through further studies, as you say. 


  • Agree x 4

#158 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 07 December 2019 - 03:51 PM

External ALA goes into the core of the "poweplant" the mitocondria (that are really similar to ancient bacteria) and sinergizing with different chemical environment can stabilize -on a permanent basis- the expression of the mitocondrial Crebs Cycle as it is: I say in two different directions: stabilize -maybe- in a "clean" direction perhaps by losing some efficiency in ATP production like a gasoline EURO 6 car -that extend rodent life- or in a "dirty" -but more efficient- direction that doesnt extend rodent life like a gasoline EURO 2 car but increase our chances of reproduction before some natural enemy maybe eats us...

 

Lipoic acid is an essential cofactor for mitochondrial metabolism and is synthesized de novo using intermediates from mitochondrial fatty acid synthesis type II, S-adenosylmethionine and iron-sulfur clusters. This cofactor is required for catalysis by multiple mitochondrial 2-ketoacid dehydrogenase complexes, including pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase, and branched-chain ketoacid dehydrogenase. Lipoic acid also plays a critical role in stabilizing and regulating these multi-enzyme complexes.  Many of these dehydrogenases are regulated by reactive oxygen species, mediated through the disulfide bond of the prosthetic lipoyl moiety.  Collectively, its functions explain why lipoic acid is required for cell growth, mitochondrial activity and coordination of fuel metabolism. Lipoic acid is an essential cofactor for mitochondrial metabolism and is synthesized de novo using intermediates from mitochondrial fatty acid synthesis type II, S-adenosylmethionine and iron-sulfur clusters. This cofactor is required for catalysis by multiple mitochondrial 2-ketoacid dehydrogenase complexes, including pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase, and branched-chain ketoacid dehydrogenase. Lipoic acid also plays a critical role in stabilizing and regulating these multi-enzyme complexes.  Many of these dehydrogenases are regulated by reactive oxygen species, mediated through the disulfide bond of the prosthetic lipoyl moiety.  Collectively, its functions explain why lipoic acid is required for cell growth, mitochondrial activity and coordination of fuel metabolism.

 

http://www.jbc.org/c...bc.TM117.000259

 

MAYBE WE SHOUD STUDY WAYS TO EXTEND (and make cleaner) BACTERIAL LIFE.... AND THEN APPLY THEM TO OUR MITOCONDRIA....


Edited by HBRU, 07 December 2019 - 04:09 PM.


#159 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 07 December 2019 - 06:14 PM

Pity I can only see the abstract, but yes it's a pretty crazy result. Don't know of any replication but assuming it's true - my guess would be ALA effects on the insulin receptor. Easy to see how ALA could sabotage DR as antioxidants have been known to deactivate autophagy (as it requires a little ROS). Much harder to explain how ALA could preserve the benefits of DR even after normal feeding resumed. But perhaps as you say it's something to do with preserving the optimisation of mitochondria.

Might it be possible to combine intermittent fasting with ALA during the refeeding period to maximise the fasting benefits?

#160 Fafner55

  • Guest
  • 300 posts
  • 298
  • Location:USA
  • NO

Posted 07 December 2019 - 06:25 PM

Pity I can only see the abstract, 

 

The paper can be found on https://sci-hub.tw/

 

"Dietary lipoic acid supplementation can mimic or block the effect of dietary restriction on life span" https://doi.org/10.1...mad.2008.04.004



#161 JamesPaul

  • Guest
  • 87 posts
  • 40
  • Location:Florida
  • NO

Posted 08 December 2019 - 04:38 AM

In case someone is not aware, Torrent Pharmaceuticals has recalled losartan due to apparent contamination with a trace amount of NDEA.

 

https://www.fda.gov/...ium-tablets-usp

 



#162 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 08 December 2019 - 07:33 AM

Easy to see how ALA could sabotage DR as antioxidants have been known to deactivate autophagy (as it requires a little ROS). Much harder to explain how ALA could preserve the benefits of DR even after normal feeding resumed. But perhaps as you say it's something to do with preserving the optimisation of mitochondria.

 

Actually ALA is an antioxidant....

I think this antioxidants have two facese a bit like dr Jekyll and mr Hyde... and specially ALA (that I dont use)....

 

http://morelife.org/...ers/7649494.pdf

 

Also autophagy (in older age....) doesnt seem to be such a game changer as it works wrongly... Maybe by breacking down tooo many usefully proteins along with bad ones.... ?!?

Richly explained. “Autophagy is nearly always thought of as beneficial even if it is barely working. We instead show that there are severe negative consequences when it breaks down and then you are better off bypassing it all together. It is classic AP: in young worms, autophagy is working properly and is essential to reach maturity, but after reproduction it starts to malfunction causing the worms to age,” he continued.

https://www.lifecode...rease-lifespan/

 

I think that focusing on ciclic senolitic protocols, telomeres extension protocols and having antioxidants is better than focusing on autophagy....

Neurons are maybe something we cannot touch too much (either by autophagy or senolitics)... that's why I suggest is maybe better using Pravastatin instead of other statins....

 

 

 


Edited by HBRU, 08 December 2019 - 07:54 AM.


#163 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 08 December 2019 - 08:42 AM

I think the key is TO BE SELECTIVE.... autophagy is not so much a selective process... it is a DIGESTIVE process

also inducing autophagy in already senesent cells may prolonging their little usefull (or damaging) life.... ?

beeing them the most selfish also they can stop producing for the body and concentrate on selfsurvival... ?!

so maybe eating little food (CR) may lead to the surviving of the most inefficient (but prone to survive) cells........ the one that we want to kill by senolitc approach....?!?

 

NO inhibits autophagy but leads to apoptosis.... ROS leads to autophagy.... so having a lot of Iron should be usefull for life extension ? it seems not.....

 

So I take Citrulline and Astaxantine... ;)

I dont do CR.... or maybe I can do it for some time, maybe one year (to maximize mithocondria efficiency) then STOP with an ALA course to fix the mithocondria efficiency....

https://www.ncbi.nlm...les/PMC6073798/

 

https://www.oatext.c...metabolites.php

 

 

 


Edited by HBRU, 08 December 2019 - 08:56 AM.


#164 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 08 December 2019 - 09:26 AM

It seems like that autophagy doesnt kills cancer cells but kind of "re-educate" cancer cells to be not cancerous (forever ?).... (a conservative, saving approach)

Autophagy doesnt kills senesent cells but sure cannot "re-educate" them to be younger and more efficient... or ?!? Yes sure CR help mithocondria learn to be more efficient (so to produce less harmfull substances by operating).... but CR doesnt help the cells do the same as mitocondria (besides autophagy).... So better point on apopotois than autophagy IMHO....

 

https://www.scienced...90123131706.htm

 

 

Considering what above Atorvastatin is better than Pravastatin for senolitics and anticancer effects..... (Pravastatin a more conservative, saving approach)

 

"Here we observed that atorvastatin (AS) can increase intracellular reactive oxygen species (ROS) and induce necrotic cell death and autophagy in NIT-1 cells, whereas pravastatin (PS) does not cause ROS and cell death but also induces autophagy."

 

https://www.hindawi....r/2016/1828071/

 

 

 


Edited by HBRU, 08 December 2019 - 09:34 AM.


#165 Andey

  • Guest
  • 673 posts
  • 203
  • Location:Kiev, Ukraine

Posted 08 December 2019 - 10:32 AM

I think we need a separate thread for this stuff as it only tangentially relates to the QuestForLife`s extend telomeres protocol.


  • Agree x 1

#166 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 08 December 2019 - 12:24 PM

I think we need a separate thread for this stuff as it only tangentially relates to the QuestForLife`s extend telomeres protocol.

Agreed this has all become very random.

The autophagy stuff is vaguely related. In brief answering a few points raised:
Autophagy can lead to senescence in the presence of telomere damage. Hard to say if this is good or bad. I started a another thread about this in the telomere section.
I doubt statins induce autophagy. I looked into this before and there is contradictory evidence. I have never seen any evidence they are senolytic (although I've seen arguments autophagy can trigger senolysis as well as senescence).
And the possible diabetes problem from long term statin use shouldn't have any impact on short term use. I expect generating too many progenitors at the expense of older but still working somatic cells can reduce the effectiveness of an organ in the short term. I've noticed a worse/stronger effect from alcohol during the protocol.

Regarding the losartan recall. I'm aware. Contamination was very low, but still concerning. Anything made in India right now is probably suspect if used all the time. A short protocol at low dose repeated 3 or 4 times a year should not cause any issues.

Edited by QuestforLife, 08 December 2019 - 12:32 PM.

  • Informative x 1

#167 HBRU

  • Guest
  • 167 posts
  • 45
  • Location:Italy
  • NO

Posted 08 December 2019 - 12:51 PM

Thanks and sorry for the confusion. Hope having given some good ideas. In short in my opinion CR (and so forced autphagia trough MTOR inhibition) mostly is good for mitocondria but not for cells as it may lead to senesent -just able to survive- cells that would be better be died. Will not add more. Thanks again.

#168 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 12 December 2019 - 11:25 AM

After not getting any results from my statin skin cream, I’ve looked again at the concentrations used in the paper where they looked at improved skin healing in rabbits. Following their results I realised I needed between 2% and 10% [doi: 10.1097/GOX.0000000000001294]. I had to use my entire stock of 1g simvastatin. I tried for a 4% mix (1 of sim in 25g atopalm). To my horror after crushing the many, many 20mg pills and sieving them to remove the casing they weighed 16g! So there is a lot of filler. If I repeat this experiment in future I will have to go for 80mg pills to minimise the filling. 16g of simvastatin and filler in 25g atopalm made a sticky paste. I had to put in another 15 g of atopalm to dissolve it all satisfactorily. I am not happy with all the filler in there (ZOCOR for oral administration contains the following inactive ingredients: ascorbic acid, citric acid, hydroxypropyl cellulose, hypromellose, iron oxides, lactose, magnesium stearate, microcrystalline cellulose, starch, talc, and titanium dioxide) but at least I've got a 2.5% mix (1g simvastatin in 40g atopalm). I tried it out last night and it absorbs well into the skin of my face without giving any discomfort. We'll see if I get any results this time.


  • Informative x 2

#169 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 17 December 2019 - 09:59 AM

Telomere length from Life Length

 

Life Length are the premier telomere measurement company in the world, established by Maria Blasco. They give a mean, median and measurement of short telomeres. They can also supply a histogram of all telomeres measured on request. It requires a venous blood draw.

 

Since the last test my age increased from 39.7 to 41
Mean telomere length improved from 13.2 to 13.4 kbp
Median telomere length improved from 11.4 to 11.7 kbp. Taking account of my increasing chronological age this represents moving from the 55 to the 74 percentile compared to other individuals in their database.
The median TL is used to calculate biological age; my estimated age improved from 38.2 (chronological 39.7) years to 37.1 (chronological 41) years. This constitutes an age reversal (change in the delta) of 3.9-1.5=2.4 years. I regard this as being a very small change.

 

More interesting is the change in the shortest telomeres. This is represented by drawing a line at the telomere length of the shortest 20% of all telomeres measured (see histogram). This improved from 6.2 to 7.1 kbp. This moved me from the 36 percentile to the 79 percentile. This is particularly pleasing for reasons I’ll discuss below.

 

Life Length supplied me with a histogram of my 2019 measurements but unfortunately the scale was different to the histogram they supplied in 2018 meaning they can’t be visually compared in a meaningful way, so I’ve asked them to re-scale and will post a comparison here when they respond. In the meantime we can make certain deductions from the fact the shortest 20% TL moved up by 900bp, the median TL moved up by 300bp and the mean TL moved up by 200bp. This tells me that the hump of the slope has shifted to the right, making a large difference at the lower end but falling off very quickly afterwards, meaning the median and mean change much less than the shortest 20%.

 

Interventions since 2018: I deliberately left it a month between the end of my last SES (Statin-Epitalon-Sartan) 20 day cycle and the Life Length test to avoid a distorting boost in leukocyte telomere length that would not reflect the overall health of the progenitor and stem cell pool that supplies them. What I did not plan was a week away in November in a rubbish hotel, with reduced sleep, bad food and getting the drunkest I’ve been in 10 years, resulting in a bad hangover the day before I took the test. I am confident therefore that my telomere length improvements from 2018 to 2019 are robust and not an artefact of a transitory intervention.


  • Informative x 1
  • WellResearched x 1

#170 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 17 December 2019 - 10:47 AM

Telomeres are not passive in ageing

This post is prompted by comments that telomeres are passive in ageing, rather than a primary driver.

Telomers shorten progressively with divisions and this changes gene expression. This is an intentional program to limit the lifetime of a cell line, necessitating their replacement from a stem cell source. It’s like clockwork – white blood cells, fat cells, muscle cells, skin cells – they all follow this pattern, with a small stem cell pool expanding out into a larger, more differentiated progenitor pool, and this feeding an even larger pool of somatic cells doing the job of keeping the body alive. For each compartment, the progenitor pool is an appropriate size for the rate of replacement needed by the tissue it serves [10.1038/ncomms2602]. White blood cells need rapid replacement; skin, fat and muscle have a smaller pool but with longer telomeres giving an equal regenerative capacity overall. Michael West has shown that no stem cells have significant telomerase activity after the embryonic-fetal transition [10.2217/rme-2019-0062]; they must have some telomerase activity in my view, but insufficient to prevent shortening.

The telomere length of your white blood cells (for example) can go up and down depending on their relative attrition and replacement rate. This is behind claims of meditation and exercise or clean eating ‘making you younger’. It does not make you younger. It does slow ageing. It alters the balance of replacement, but the stem cells are still ageing via telomere shortening and this will eventually lead to a reduction in replacement rate of the somatic cells.

A similar but reciprocal argument holds for DNA methylation, which builds up in long lived somatic cells, but is largely reset in cells replaced regularly from stem cell pools. Hence you could reduce your ‘methylation age’ by accelerating ageing with faster replacement or increase your ‘methylation age’ by slowing replacement but keeping more stem cells for the future. Neither is optimal or what we want.

In practice I advocate cyclical telomerase expression combined with an upward pressure on somatic cells to de-differentiate some portion of them back to progenitors. This allows for epigenetic control whilst you extend telomeres (and it makes it easier to extend telomeres as well). In the longer periods between cycles cells can differentiate normally. It is my belief that dividing cells and those with the shortest telomeres benefit the most from telomerase, so stem cells are likely to benefit least, but this also means that over multiple cycles they should gradually be rejuvenated whilst the larger effects on somatic and some progenitors come out in the wash, or at best reduce the demand on stem cells (which might act as a negative feedback loop in the case of continuous telomerase activity).


  • like x 1

#171 Oakman

  • Location:CO

Posted 17 December 2019 - 01:45 PM

Interventions since 2018: I deliberately left it a month between the end of my last SES (Statin-Epitalon-Sartan) 20 day cycle and the Life Length test to avoid a distorting boost in leukocyte telomere length that would not reflect the overall health of the progenitor and stem cell pool that supplies them. What I did not plan was a week away in November in a rubbish hotel, with reduced sleep, bad food and getting the drunkest I’ve been in 10 years, resulting in a bad hangover the day before I took the test. I am confident therefore that my telomere length improvements from 2018 to 2019 are robust and not an artefact of a transitory intervention.

 

You make it sound like telomere length is something that can be affected by transitory events, over a short period of time. Isn't telomere length the result of gradual changes over time? How is it possible for a drunk night and a short intervention (days) able to change anything to any significant degree or were you just rationalizing what you want to believe?



#172 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 17 December 2019 - 03:05 PM

You make it sound like telomere length is something that can be affected by transitory events, over a short period of time. Isn't telomere length the result of gradual changes over time? How is it possible for a drunk night and a short intervention (days) able to change anything to any significant degree or were you just rationalizing what you want to believe?

 

Below is a detailed explanation I planned to post in any case, but it covers your question. In summary, white blood cells turnover every 6 days or so, and are replaced from the progenitor pool. Therefore various positive or negative lifestyle interventions can make your leukocyte telomeres get longer or shorter, but really all you're doing is varying their rate of attrition and/or replacement. 

 

Here is the more detailed answer.

 

Telomeres shorten progressively with divisions and this changes gene expression. This is an intentional program to limit the lifetime of a cell line, necessitating their replacement from a stem cell source. It’s like clockwork – white blood cells, fat cells, muscle cells, skin cells – they all follow this pattern, with a small stem cell pool expanding out into a larger, more differentiated progenitor pool, and this feeding an even larger pool of somatic cells doing the job of keeping the body alive. For each compartment, the progenitor pool is an appropriate size for the rate of replacement needed by the tissue it serves [10.1038/ncomms2602]. White blood cells need rapid replacement; skin, fat and muscle have a smaller pool but with longer telomeres giving an equal regenerative capacity overall. Michael West has shown that no stem cells have significant telomerase activity after the embryonic-fetal transition [10.2217/rme-2019-0062]; they must have some telomerase activity in my view, but insufficient to prevent shortening.

The telomere length of your white blood cells (for example) can go up and down depending on their relative attrition and replacement rate. This is behind claims of meditation and exercise or clean eating ‘making you younger’. It does not make you younger. It does slow ageing if sustained for long periods by altering the balance of replacement, but the stem cells are still ageing via telomere shortening and this will eventually lead to a reduction in replacement rate of the somatic cells.

A similar but reciprocal argument holds for DNA methylation, which builds up in long lived somatic cells, but is largely reset in cells replaced regularly from stem cell pools. Hence you could reduce your ‘methylation age’ by accelerating ageing with faster replacement or increase your ‘methylation age’ by slowing replacement but keeping more stem cells for the future. Neither is optimal or what we want.

In practice I advocate cyclical telomerase expression combined with an upward pressure on somatic cells to de-differentiate some portion of them back to progenitors. This allows for epigenetic control whilst you extend telomeres (and it makes it easier to extend telomeres as well). In the longer periods between cycles cells can differentiate normally. It is my belief that dividing cells and those with the shortest telomeres benefit the most from telomerase, so stem cells are likely to benefit least, but this also means that over multiple cycles they should gradually be rejuvenated whilst the larger effects on somatic and some progenitors come out in the wash, or at best reduce the demand on stem cells (which might act as a negative feedback loop in the case of continuous telomerase activity).


  • Good Point x 2

#173 dlewis1453

  • Member
  • 151 posts
  • 45
  • Location:USA
  • NO

Posted 17 December 2019 - 04:15 PM

 

In practice I advocate cyclical telomerase expression combined with an upward pressure on somatic cells to de-differentiate some portion of them back to progenitors. This allows for epigenetic control whilst you extend telomeres (and it makes it easier to extend telomeres as well). In the longer periods between cycles cells can differentiate normally. It is my belief that dividing cells and those with the shortest telomeres benefit the most from telomerase, so stem cells are likely to benefit least, but this also means that over multiple cycles they should gradually be rejuvenated whilst the larger effects on somatic and some progenitors come out in the wash, or at best reduce the demand on stem cells (which might act as a negative feedback loop in the case of continuous telomerase activity).

 

Thanks for the thorough post QuestforLife. It sounds as if you have developed a carefully thought out protocol. Could you elaborate some more on your protocol? 



#174 Turnbuckle

  • Location:USA
  • NO

Posted 17 December 2019 - 05:06 PM

 

In practice I advocate cyclical telomerase expression combined with an upward pressure on somatic cells to de-differentiate some portion of them back to progenitors. 

 

 

What method are you using for the part in bold?



#175 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 18 December 2019 - 09:12 AM

What method are you using for the part in bold?

 

Here is a probably more detailed answer than you wanted.

 

I use Rho kinase (ROCK for short) inhibitors. ROCK controls the cytoskeleton of the cells, and hence its shape, which has a direct bearing on its stemness. Here is one of a remarkable series of papers on what ROCK inhibitors can do in vitro.
http://stemcellres.com/content/5/2/60

We previously demonstrated that the lifespan of primary human keratinocytes could be extended indefinitely by culture in the presence of the Rho kinase (ROCK) inhibitor Y-27632. This technique has proven to be very useful in diverse areas of basic and clinical research.

Y-27632 is not available so I was able to identify statins as ROCK inhibitors, both increasing stemness in vitro;
https://journals.plo...al.pone.0120137

Importantly, we observed that simvastatin increased the percentage of a subset of smaller MSC, which also were actively proliferating. The association of MSC decreased size with increased pluripotency and the accumulating evidence that statins may prevent cellular senescence led us to hypothesize that simvastatin induces a smaller subpopulation that may have increased ability to maintain the entire pool of MSC and also to protect them from cellular senescence induced by long-term cultures/passages in vitro.

(note Klf4 is downregulated by ROCK)
And inhibiting ROCK in vivo:
https://journals.sag...323001103900630

Short-term treatment with either rosuvastatin or atorvastatin inhibits ROCK activity independent of cholesterol reduction, and improves endothelium dysfunction in patients with atherosclerosis.

Most interestingly, a low dose of Fluvastatin has significant health benefits and involves telomerase (ROCK inhibition activates telomerase)
https://www.ncbi.nlm...pubmed/26214555

We found that a low-dose combination of fluvastatin and valsartan substantially
increased telomerase activity, which significantly correlated with an improvement of endothelial
function and a decrease of inflammation/oxidative stress. These findings could lead to a new
innovative approach to arterial rejuvenation.

 

And finally users of statins are well known to have longer telomeres than age matched controls (despite the side effects of long term use)

https://www.frontier...2016.00347/full

 

These initial observations suggest atorvastatin can act as telomerase activator and potentially as effective geroprotector

 

I am in the process of identifying natural ROCK inhibitors so I may be able to use them instead of statins, but that will have to wait for a future report.


  • Informative x 3

#176 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 18 December 2019 - 09:15 AM

Thanks for the thorough post QuestforLife. It sounds as if you have developed a carefully thought out protocol. Could you elaborate some more on your protocol? 

 

My latest report from the start of November is here: https://www.longecit...e-5#entry881808

 

This gives a full explanation of the 20 day protocol.


Edited by QuestforLife, 18 December 2019 - 09:16 AM.

  • Informative x 1

#177 Turnbuckle

  • Location:USA
  • NO

Posted 18 December 2019 - 12:34 PM

My latest report from the start of November is here: https://www.longecit...e-5#entry881808

 

This gives a full explanation of the 20 day protocol.

 

 

Have you been able to reduce epigenetic age this way?



#178 QuestforLife

  • Topic Starter
  • Guest
  • 1,524 posts
  • 1,135
  • Location:UK
  • NO

Posted 18 December 2019 - 03:03 PM

Have you been able to reduce epigenetic age this way?

 

My recent blood panel (Nov 2019) gives a very young PhenoAge of 30 (I am 41) and suggests a DNAm age of just under 30, though I am cautious about believing the latter until I can verify this with a direct DNAm age test. 

 

From my Results page in the member area...


PhenoAge Blood Panel

At the start of November I also had a draw for a comprehensive blood panel allowing me to calculate my PhenoAge. Thank you John G. Cramer for compiling the excel table allowing this calculation. My inputs to the table were as follows:
Albumin: 4.6 g/dL
Creatinine: 1.007 mg/dL
Glucose: 73.9 mg/dL
C-Reactive Protein: 1 mg/L
Lympocytes: 39.3%
Mean Cell Volume: 84.8 fL
Red Cell Distribution Width: 13.2%
Alkaline Phosphatase: 79 U/L
White Blood Cells: 5 x 10^3 cells/uL
Age: 41
This gave the following results:
MortScore: 0.008
Phenotypic Age: 30.04
Est. DNAm Age: 29.84
Est D MScore: 0.008

 

My last DNAm age in April 2019 gave an age of 38 (delta with chronological age of 2.4 years) compared to my result in July 2018, for which my chronological age and methylation age did not vary significantly. This tells me that low dose rapamycin taken weekly for 2 years before 2018 did not reduce my methylation age, but something else I did in 2018/2019 (probably this protocol) did. 

 

I will continue to experiment with this protocol and continue to get tests done in 2020 and hopefully I will be able to report more interesting (and conclusive) results. 


  • Informative x 3

#179 dlewis1453

  • Member
  • 151 posts
  • 45
  • Location:USA
  • NO

Posted 18 December 2019 - 03:22 PM

 

 

My last DNAm age in April 2019 gave an age of 38 (delta with chronological age of 2.4 years) compared to my result in July 2018, for which my chronological age and methylation age did not vary significantly. This tells me that low dose rapamycin taken weekly for 2 years before 2018 did not reduce my methylation age, but something else I did in 2018/2019 (probably this protocol) did. 

 

 

 

FYI - I read in an interview with Dr. Alan Green (the rapamycin doctor) that he has patients that initially reduce epigenetic age by 6 to 12 years after a year on Rapamycin. Epigenetic aging is then slowed going forward. However, from his interviews I gathered that this benefit, and the benefits of mtor suppression by Rapamycin generally, do not kick in until sometime is in the 40s at the earliest. The cases of large epigenetic age regression seem to have all happened in elderly people. 

 

It's possible that you are simply too young to get substantial benefit from Rapamycin right now. However, it may begin to make more of a difference for your longevity trajectory in your mid 40s. Although you seem very healthy and you have already done so many longevity interventions, so your Rapamycin benefit window may be pushed back into your 50s. Its hard to say. 



#180 Turnbuckle

  • Location:USA
  • NO

Posted 18 December 2019 - 03:25 PM

My recent blood panel (Nov 2019) gives a very young PhenoAge of 30 (I am 41) and suggests a DNAm age of just under 30, though I am cautious about believing the latter until I can verify this with a direct DNAm age test. 

 

From my Results page in the member area...

 

 

My last DNAm age in April 2019 gave an age of 38 (delta with chronological age of 2.4 years) compared to my result in July 2018, for which my chronological age and methylation age did not vary significantly. This tells me that low dose rapamycin taken weekly for 2 years before 2018 did not reduce my methylation age, but something else I did in 2018/2019 (probably this protocol) did. 

 

I will continue to experiment with this protocol and continue to get tests done in 2020 and hopefully I will be able to report more interesting (and conclusive) results. 

 

 

If you really are de-differentiating somatic cells, that should definitely lower your epigenetic age, no matter how old you are chronologically. It would be a fantastic breakthrough.

 

BTW, I see that one ROCK inhibitor--fasudil--promotes gliogenesis of neural stem cells, which isn't a good thing.

 

Exposure to the ROCK inhibitor fasudil promotes gliogenesis of neural stem cells in vitro.


Edited by Turnbuckle, 18 December 2019 - 03:41 PM.

  • Informative x 3





Also tagged with one or more of these keywords: telomeres, nad, nampt, ampk, resveratrol, allicin, methylene blue, nmn, sirtuins, statin

0 user(s) are reading this topic

0 members, 0 guests, 0 anonymous users